1
|
Liu J, Prahl LS, Huang AZ, Hughes AJ. Measurement of adhesion and traction of cells at high yield reveals an energetic ratchet operating during nephron condensation. Proc Natl Acad Sci U S A 2024; 121:e2404586121. [PMID: 39292750 PMCID: PMC11441508 DOI: 10.1073/pnas.2404586121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/21/2024] [Indexed: 09/20/2024] Open
Abstract
Developmental biology-inspired strategies for tissue-building have extraordinary promise for regenerative medicine, spurring interest in the relationship between cell biophysical properties and morphological transitions. However, mapping gene or protein expression data to cell biophysical properties to physical morphogenesis remains challenging with current techniques. Here, we present multiplexed adhesion and traction of cells at high yield (MATCHY). MATCHY advances the multiplexing and throughput capabilities of existing traction force and cell-cell adhesion assays using microfabrication and a semiautomated computation scheme with machine learning-driven cell segmentation. Both biophysical assays are coupled with serial downstream immunofluorescence to extract cell type/signaling state information. MATCHY is especially suited to complex primary tissue-, organoid-, or biopsy-derived cell mixtures since it does not rely on a priori knowledge of cell surface markers, cell sorting, or use of lineage-specific reporter animals. We first validate MATCHY on canine kidney epithelial cells engineered for rearranged during transfection (RET) tyrosine kinase expression and quantify a relationship between downstream signaling and cell traction. We then use MATCHY to create a biophysical atlas of mouse embryonic kidney primary cells and identify distinct biophysical states along the nephron differentiation trajectory. Our data complement expression-level knowledge of adhesion molecule changes that accompany nephron differentiation with quantitative biophysical information. These data reveal an "energetic ratchet" that accounts for spatial trends in nephron progenitor cell condensation as they differentiate into early nephron structures, which we validate through agent-based computational simulation. MATCHY offers semiautomated cell biophysical characterization at >10,000-cell throughput, an advance benefiting fundamental studies and new synthetic tissue strategies for regenerative medicine.
Collapse
Affiliation(s)
- Jiageng Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA19104
| | - Louis S. Prahl
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA19104
| | - Aria Zheyuan Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA19104
| | - Alex J. Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA19104
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, PA19104
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, PA19104
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA19104
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, PA19104
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA19104
- Materials Research Science and Engineering Center, University of Pennsylvania, Philadelphia, PA19104
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
2
|
Der B, Bugacov H, Briantseva BM, McMahon AP. Cadherin adhesion complexes direct cell aggregation in the epithelial transition of Wnt-induced nephron progenitor cells. Development 2024; 151:dev202303. [PMID: 39344436 PMCID: PMC11463967 DOI: 10.1242/dev.202303] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 06/24/2024] [Indexed: 10/01/2024]
Abstract
In the developing mammalian kidney, nephron formation is initiated by a subset of nephron progenitor cells (NPCs). Wnt input activates a β-catenin (Ctnnb1)-driven, transcriptional nephrogenic program and the mesenchymal to epithelial transition (MET) of NPCs. Using an in vitro mouse NPC culture model, we observed that activation of the Wnt pathway results in the aggregation of induced NPCs, which is an initiating step in the MET program. Genetic removal showed aggregation was dependent on β-catenin. Modulating extracellular Ca2+ levels showed cell-cell contacts were Ca2+ dependent, suggesting a role for cadherin (Cdh)-directed cell adhesion. Molecular analysis identified Cdh2, Cdh4 and Cdh11 in NPCs, and the β-catenin directed upregulation of Cdh3 and Cdh4 accompanying the MET of induced NPCs. Mutational analysis of β-catenin supported a role for a Lef/Tcf-β-catenin-mediated transcriptional response in the cell aggregation process. Genetic removal of all four cadherins, and independent removal of α-catenin or of β-catenin-α-catenin interactions, abolished aggregation, but not the inductive response to Wnt pathway activation. These findings, and data in an accompanying article highlight the role of β-catenin in linking transcriptional programs to the morphogenesis of NPCs in mammalian nephrogenesis.
Collapse
Affiliation(s)
- Balint Der
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles 90033, USA
- Department of Urology, Faculty of Medicine, Semmelweis University, Budapest 1082, Hungary
- Institute of Translational Medicine, Faculty of Medicine, Semmelweis University, Budapest 1094, Hungary
| | - Helena Bugacov
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles 90033, USA
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bohdana-Myroslava Briantseva
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles 90033, USA
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles 90033, USA
| |
Collapse
|
3
|
Song L, Li Q, Xia L, Sahay AE, Qiu Q, Li Y, Li H, Sasaki K, Susztak K, Wu H, Wan L. Single-cell multiomics reveals ENL mutation perturbs kidney developmental trajectory by rewiring gene regulatory landscape. Nat Commun 2024; 15:5937. [PMID: 39009564 PMCID: PMC11250843 DOI: 10.1038/s41467-024-50171-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 07/02/2024] [Indexed: 07/17/2024] Open
Abstract
How disruptions to normal cell differentiation link to tumorigenesis remains incompletely understood. Wilms tumor, an embryonal tumor associated with disrupted organogenesis, often harbors mutations in epigenetic regulators, but their role in kidney development remains unexplored. Here, we show at single-cell resolution that a Wilms tumor-associated mutation in the histone acetylation reader ENL disrupts kidney differentiation in mice by rewiring the gene regulatory landscape. Mutant ENL promotes nephron progenitor commitment while restricting their differentiation by dysregulating transcription factors such as Hox clusters. It also induces abnormal progenitors that lose kidney-associated chromatin identity. Furthermore, mutant ENL alters the transcriptome and chromatin accessibility of stromal progenitors, resulting in hyperactivation of Wnt signaling. The impacts of mutant ENL on both nephron and stroma lineages lead to profound kidney developmental defects and postnatal mortality in mice. Notably, a small molecule inhibiting mutant ENL's histone acetylation binding activity largely reverses these defects. This study provides insights into how mutations in epigenetic regulators disrupt kidney development and suggests a potential therapeutic approach.
Collapse
Affiliation(s)
- Lele Song
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qinglan Li
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Lingbo Xia
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of the School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Arushi Eesha Sahay
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Qi Qiu
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yuanyuan Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Haitao Li
- MOE Key Laboratory of Protein Sciences, Beijing Frontier Research Center for Biological Structure, School of Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China
| | - Kotaro Sasaki
- Department of Biomedical Sciences, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA, 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Katalin Susztak
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104, USA
- Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Hao Wu
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Liling Wan
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
4
|
Park A, Kim S, Yu J, Son D, Kim K, Koh E, Park K. Cadherin-6 is a novel mediator for the migration of mesenchymal stem cells to glioblastoma cells in response to stromal cell-derived factor-1. FEBS Open Bio 2024; 14:1192-1204. [PMID: 38719785 PMCID: PMC11216932 DOI: 10.1002/2211-5463.13815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/03/2024] [Accepted: 04/29/2024] [Indexed: 07/03/2024] Open
Abstract
Glioblastoma recruits various nontransformed cells from distant tissues. Although bone marrow-derived mesenchymal stem cells (MSCs) have been observed migrating to glioblastoma, the underlying mechanism driving MSC migration toward glioblastoma remains unclear. Tumor vascularity is critical in the context of recurrent glioblastoma and is closely linked to the expression of stromal cell-derived factor-1 (SDF-1). We demonstrated that cadherin-6 mediated MSC migration both toward SDF-1 and toward glioblastoma cells. Cadherin-6 knockdown resulted in the downregulation of MSCs capacity to migrate in response to SDF-1. Furthermore, MSCs with cadherin-6 knockdown exhibited impaired migration in response to conditioned media derived from glioblastoma cell lines (U87 and U373) expressing SDF-1, thus simulating the glioblastoma microenvironment. Moreover, MSCs enhanced the vasculogenic capacity of U87 cells without increasing the proliferation, cancer stem cell characteristics, or migration of U87. These results suggest that the current strategy of utilizing MSCs as carriers for antiglioblastoma drugs requires careful examination. Furthermore, cadherin-6 may represent a novel potential target for controlling the recruitment of MSCs toward glioblastoma.
Collapse
Affiliation(s)
- Aran Park
- Graduate School of BiotechnologyKyung Hee UniversityYonginKorea
| | - Seung‐Eun Kim
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Jinyeong Yu
- Industry‐Academic Cooperation FoundationKyung Hee UniversitySeoulKorea
| | - Donghyun Son
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
| | - Kyung‐Sup Kim
- Department of Biochemistry and Molecular Biology, College of MedicineYonsei UniversitySeoulKorea
| | - Eunjin Koh
- Department of Biochemistry and Molecular Biology, College of MedicineYonsei UniversitySeoulKorea
| | - Ki‐Sook Park
- Department of Biomedical Science and Technology, Graduate SchoolKyung Hee UniversitySeoulKorea
- East‐West Medical Research InstituteKyung Hee UniversitySeoulKorea
| |
Collapse
|
5
|
Song L, Li Q, Xia L, Sahay A, Qiu Q, Li Y, Li H, Sasaki K, Susztak K, Wu H, Wan L. Single-Cell multiomics reveals ENL mutation perturbs kidney developmental trajectory by rewiring gene regulatory landscape. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.591709. [PMID: 38766219 PMCID: PMC11100752 DOI: 10.1101/2024.05.09.591709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Cell differentiation during organogenesis relies on precise epigenetic and transcriptional control. Disruptions to this regulation can result in developmental abnormalities and malignancies, yet the underlying mechanisms are not well understood. Wilms tumors, a type of embryonal tumor closely linked to disrupted organogenesis, harbor mutations in epigenetic regulators in 30-50% of cases. However, the role of these regulators in kidney development and pathogenesis remains unexplored. By integrating mouse modeling, histological characterizations, and single-cell transcriptomics and chromatin accessibility profiling, we show that a Wilms tumor-associated mutation in the chromatin reader protein ENL disrupts kidney development trajectory by rewiring the gene regulatory landscape. Specifically, the mutant ENL promotes the commitment of nephron progenitors while simultaneously restricting their differentiation by dysregulating key transcription factor regulons, particularly the HOX clusters. It also induces the emergence of abnormal progenitor cells that lose their chromatin identity associated with kidney specification. Furthermore, the mutant ENL might modulate stroma-nephron interactions via paracrine Wnt signaling. These multifaceted effects caused by the mutation result in severe developmental defects in the kidney and early postnatal mortality in mice. Notably, transient inhibition of the histone acetylation binding activity of mutant ENL with a small molecule displaces transcriptional condensates formed by mutant ENL from target genes, abolishes its gene activation function, and restores developmental defects in mice. This work provides new insights into how mutations in epigenetic regulators can alter the gene regulatory landscape to disrupt kidney developmental programs at single-cell resolution in vivo . It also offers a proof-of-concept for the use of epigenetics-targeted agents to rectify developmental defects.
Collapse
|
6
|
Chambers BE, Weaver NE, Lara CM, Nguyen TK, Wingert RA. (Zebra)fishing for nephrogenesis genes. Tissue Barriers 2024; 12:2219605. [PMID: 37254823 PMCID: PMC11042071 DOI: 10.1080/21688370.2023.2219605] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/14/2023] [Indexed: 06/01/2023] Open
Abstract
Kidney disease is a devastating condition affecting millions of people worldwide, where over 100,000 patients in the United States alone remain waiting for a lifesaving organ transplant. Concomitant with a surge in personalized medicine, single-gene mutations, and polygenic risk alleles have been brought to the forefront as core causes of a spectrum of renal disorders. With the increasing prevalence of kidney disease, it is imperative to make substantial strides in the field of kidney genetics. Nephrons, the core functional units of the kidney, are epithelial tubules that act as gatekeepers of body homeostasis by absorbing and secreting ions, water, and small molecules to filter the blood. Each nephron contains a series of proximal and distal segments with explicit metabolic functions. The embryonic zebrafish provides an ideal platform to systematically dissect the genetic cues governing kidney development. Here, we review the use of zebrafish to discover nephrogenesis genes.
Collapse
Affiliation(s)
- Brooke E. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Nicole E. Weaver
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Caroline M. Lara
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Thanh Khoa Nguyen
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, Warren Center for Drug Discovery, University of Notre Dame, Notre Dame, Indiana (IN), USA
| |
Collapse
|
7
|
Suzuki H, Nagase S, Saito C, Takatsuka A, Nagata M, Honda K, Kaneda Y, Nishiya Y, Honda T, Ishizaka T, Nakamura K, Nakada T, Abe Y, Agatsuma T. Raludotatug Deruxtecan, a CDH6-Targeting Antibody-Drug Conjugate with a DNA Topoisomerase I Inhibitor DXd, Is Efficacious in Human Ovarian and Kidney Cancer Models. Mol Cancer Ther 2024; 23:257-271. [PMID: 38205802 PMCID: PMC10911705 DOI: 10.1158/1535-7163.mct-23-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/28/2023] [Accepted: 01/05/2024] [Indexed: 01/12/2024]
Abstract
Cadherin-6 (CDH6) is expressed in several cancer types, but no CDH6-targeted therapy is currently clinically available. Here, we generated raludotatug deruxtecan (R-DXd; DS-6000), a novel CDH6-targeting antibody-drug conjugate with a potent DNA topoisomerase I inhibitor, and evaluated its properties, pharmacologic activities, and safety profile. In vitro pharmacologic activities and the mechanisms of action of R-DXd were assessed in serous-type ovarian cancer and renal cell carcinoma cell lines. In vivo pharmacologic activities were evaluated with several human cancer cell lines and patient-derived xenograft mouse models. The safety profile in cynomolgus monkeys was also assessed. R-DXd exhibited CDH6 expression-dependent cell growth-inhibitory activity and induced tumor regression in xenograft models. In this process, R-DXd specifically bound to CDH6, was internalized into cancer cells, and then translocated to the lysosome. The DXd released from R-DXd induced the phosphorylation of Chk1, a DNA damage marker, and cleaved caspase-3, an apoptosis marker, in cancer cells. It was also confirmed that the DXd payload had a bystander effect, passing through the cell membrane and impacting surrounding cells. The safety profile of R-DXd was favorable and the highest non-severely toxic dose was 30 mg/kg in cynomolgus monkeys. R-DXd demonstrated potent antitumor activity against CDH6-expressing tumors in mice and an acceptable safety profile in monkeys. These findings indicate the potential of R-DXd as a new treatment option for patients with CDH6-expressing serous-type ovarian cancer and renal cell carcinoma in a clinical setting.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yuki Abe
- Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | |
Collapse
|
8
|
Farouk SM, Basha WAA, Emam MA, Metwally E. Differential expression of epithelial and smooth muscle lineage-specific markers of metanephros in one-humped camel foetuses. Anat Histol Embryol 2024; 53:e12985. [PMID: 37814965 DOI: 10.1111/ahe.12985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/28/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
The development of the metanephros in one-humped camels involves a complex series of interactions between epithelial and mesenchymal cells. As a result, there is a synchronized differentiation process of stromal, vascular and epithelial cell types during glomerulogenesis, angiogenesis and tubulogenesis. In the current work, the metanephros of camel foetuses were divided into four stages where kidneys from each stage were processed and immunoassayed, followed by quantitative analysis to determine target protein intensities throughout metanephrogenesis in the camel. This study demonstrated robust expression of α-smooth muscle actin (α-SMA) in the glomerular mesangium, as well as in interlobular and glomerular arterioles during the earlier stages of development. However, in the late stages, α-SMA expression became more localized around the blood capillaries in both the cortex and medulla. Strong expression of CD34 was observed in the immature glomerular and peritubular endothelial cells within the subcapsular zone, as well as in the glomerular, proximal tubular and distal tubular epithelium of stage one foetuses, although its expression gradually diminished with foetal maturation. The expression pattern of osteopontin was prominently observed in the distal convoluted tubules throughout all stages, however, no expression was detected in the proximal tubules, glomeruli and arterioles. E-cadherin was detected in the developing renal tubular epithelial cells but not in the glomeruli. In conclusion, this study reveals the spatiotemporal distribution of key proteins, including α-SMA, CD34, Osteopontin and E-cadherin, which play a crucial role in metanephrogenesis in camel foetuses.
Collapse
Affiliation(s)
- Sameh M Farouk
- Cytology and Histology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Walaa A A Basha
- Anatomy and Embryology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Mahmoud A Emam
- Histology Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Elsayed Metwally
- Cytology and Histology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
9
|
Guo M, Shen D, Su Y, Xu J, Zhao S, Zhang W, Wang Y, Jiang W, Wang J, Geng X, Ding X, Xu X. Syndecan-1 shedding destroys epithelial adherens junctions through STAT3 after renal ischemia/reperfusion injury. iScience 2023; 26:108211. [PMID: 37942007 PMCID: PMC10628745 DOI: 10.1016/j.isci.2023.108211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023] Open
Abstract
Adherens junctions between tubular epithelial cells are disrupted in renal ischemia/reperfusion (I/R) injury. Syndecan-1 (SDC-1) is involved in maintaining cell morphology. We aimed to study the role of SDC-1 shedding induced by renal I/R in the destruction of intracellular adherens junctions. We found that SDC-1 shedding was increased while the expression of E-cadherin was decreased. This observation was accompanied by the activation of STAT3 in the kidneys. Inhibiting the shedding of SDC-1 induced by I/R could alleviate this effect. Mild renal I/R could induce more severe renal injury, lower E-cadherin expression, damaged cell junctions, and activated STAT3 in knockout mice with the tubule-specific deletion of SDC-1 mice. The results in vitro were consistent with those in vivo. Inhibiting the shedding of SDC-1 could alleviate the decreased expression of E-cadherin and damage of cell adherens junctions through inhibiting the activation of STAT3 during ischemic acute kidney injury.
Collapse
Affiliation(s)
- Man Guo
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Daoqi Shen
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Yiqi Su
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Jiarui Xu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Shuan Zhao
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Weidong Zhang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Yaqiong Wang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Wuhua Jiang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Jialin Wang
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Xuemei Geng
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Xiaoqiang Ding
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| | - Xialian Xu
- Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Medical Center of Kidney Disease, Shanghai, China
- Shanghai Institute of Kidney and Dialysis, Shanghai, China
- Key Laboratory of Kidney and Blood Purification, Shanghai, China
- Hemodialysis quality control center of Shanghai, Shanghai, China
| |
Collapse
|
10
|
Der B, Bugacov H, Briantseva BM, McMahon AP. Cadherin Adhesion Complexes Direct Cell Aggregation in the Epithelial Transition of Wnt-Induced Nephron Progenitor Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.27.555021. [PMID: 38654822 PMCID: PMC11037868 DOI: 10.1101/2023.08.27.555021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
In the developing mammalian kidney, nephron formation is initiated by a subset of nephron progenitor cells (NPCs). Wnt input activates a β-catenin ( Ctnnb1 )-driven, transcriptional nephrogenic program. In conjunction, induced mesenchymal NPCs transition through a pre-tubular aggregate to an epithelial renal vesicle, the precursor for each nephron. How this critical mesenchymal-to-epithelial transition (MET) is regulated is unclear. In an in vitro mouse NPC culture model, activation of the Wnt pathway results in the aggregation of induced NPCs into closely-packed, cell clusters. Genetic removal of β-catenin resulted in a failure of both Wnt pathway-directed transcriptional activation and the formation of aggregated cell clusters. Modulating extracellular Ca 2+ levels showed cell-cell contacts were Ca 2+ -dependent, suggesting a role for cadherin (Cdh)-directed cell adhesion. Molecular analysis identified Cdh2 , Cdh4 and Cdh11 in uninduced NPCs and the up-regulation of Cdh3 and Cdh4 accompanying the Wnt pathway-induced MET. Genetic removal of all four cadherins, and independent removal of α-catenin, which couples Cdh-β-catenin membrane complexes to the actin cytoskeleton, abolished cell aggregation in response to Wnt pathway activation. However, the β-catenin driven inductive transcriptional program was unaltered. Together with the accompanying paper (Bugacov et al ., submitted), these data demonstrate that distinct cellular activities of β-catenin - transcriptional regulation and cell adhesion - combine in the mammalian kidney programs generating differentiated epithelial nephron precursors from mesenchymal nephron progenitors. Summary statement Our study highlights the role of Wnt-β-catenin pathway regulation of cadherin-mediated cell adhesion in the mesenchymal to epithelial transition of induced nephron progenitor cells.
Collapse
|
11
|
Banan Sadeghian R, Ueno R, Takata Y, Kawakami A, Ma C, Araoka T, Takasato M, Yokokawa R. Cells sorted off hiPSC-derived kidney organoids coupled with immortalized cells reliably model the proximal tubule. Commun Biol 2023; 6:483. [PMID: 37142732 PMCID: PMC10160057 DOI: 10.1038/s42003-023-04862-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Of late, numerous microphysiological systems have been employed to model the renal proximal tubule. Yet there is lack of research on refining the functions of the proximal tubule epithelial layer-selective filtration and reabsorption. In this report, pseudo proximal tubule cells extracted from human-induced pluripotent stem cell-derived kidney organoids are combined and cultured with immortalized proximal tubule cells. It is shown that the cocultured tissue is an impervious epithelium that offers improved levels of certain transporters, extracellular matrix proteins collagen and laminin, and superior glucose transport and P-glycoprotein activity. mRNA expression levels higher than those obtained from each cell type were detected, suggesting an anomalous synergistic crosstalk between the two. Alongside, the improvements in morphological characteristics and performance of the immortalized proximal tubule tissue layer exposed, upon maturation, to human umbilical vein endothelial cells are thoroughly quantified and compared. Glucose and albumin reabsorption, as well as xenobiotic efflux rates through P-glycoprotein were all improved. The data presented abreast highlight the advantages of the cocultured epithelial layer and the non-iPSC-based bilayer. The in vitro models presented herein can be helpful in personalized nephrotoxicity studies.
Collapse
Affiliation(s)
| | - Ryohei Ueno
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Yuji Takata
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Akihiko Kawakami
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Cheng Ma
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, 606-8507, Japan
| | - Minoru Takasato
- RIKEN Center for Biosystems Dynamics Research (BDR), Kobe, 650-0047, Japan
- Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto, 615-8540, Japan.
| |
Collapse
|
12
|
Cote LE, Feldman JL. Won't You be My Neighbor: How Epithelial Cells Connect Together to Build Global Tissue Polarity. Front Cell Dev Biol 2022; 10:887107. [PMID: 35800889 PMCID: PMC9253303 DOI: 10.3389/fcell.2022.887107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Epithelial tissues form continuous barriers to protect against external environments. Within these tissues, epithelial cells build environment-facing apical membranes, junction complexes that anchor neighbors together, and basolateral surfaces that face other cells. Critically, to form a continuous apical barrier, neighboring epithelial cells must align their apico-basolateral axes to create global polarity along the entire tissue. Here, we will review mechanisms of global tissue-level polarity establishment, with a focus on how neighboring epithelial cells of different origins align their apical surfaces. Epithelial cells with different developmental origins and/or that polarize at different times and places must align their respective apico-basolateral axes. Connecting different epithelial tissues into continuous sheets or tubes, termed epithelial fusion, has been most extensively studied in cases where neighboring cells initially dock at an apical-to-apical interface. However, epithelial cells can also meet basal-to-basal, posing several challenges for apical continuity. Pre-existing basement membrane between the tissues must be remodeled and/or removed, the cells involved in docking are specialized, and new cell-cell adhesions are formed. Each of these challenges can involve changes to apico-basolateral polarity of epithelial cells. This minireview highlights several in vivo examples of basal docking and how apico-basolateral polarity changes during epithelial fusion. Understanding the specific molecular mechanisms of basal docking is an area ripe for further exploration that will shed light on complex morphogenetic events that sculpt developing organisms and on the cellular mechanisms that can go awry during diseases involving the formation of cysts, fistulas, atresias, and metastases.
Collapse
|
13
|
Bradford STJ, Grimley E, Laszczyk AM, Lee PH, Patel SR, Dressler GR. Identification of Pax protein inhibitors that suppress target gene expression and cancer cell proliferation. Cell Chem Biol 2022; 29:412-422.e4. [PMID: 34822752 PMCID: PMC8934255 DOI: 10.1016/j.chembiol.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/24/2021] [Accepted: 11/02/2021] [Indexed: 11/16/2022]
Abstract
The Pax family of developmental control genes are frequently deregulated in human disease. In the kidney, Pax2 is expressed in developing nephrons but not in adult proximal and distal tubules, whereas polycystic kidney epithelia or renal cell carcinoma continues to express high levels. Pax2 reduction in mice or cell culture can slow proliferation of cystic epithelial cells or renal cancer cells. Thus, inhibition of Pax activity may be a viable, cell-type-specific therapy. We designed an unbiased, cell-based, high-throughput screen that identified triazolo pyrimidine derivatives that attenuate Pax transactivation ability. We show that BG-1 inhibits Pax2-positive cancer cell growth and target gene expression but has little effect on Pax2-negative cells. Chromatin immunoprecipitation suggests that these inhibitors prevent Pax protein interactions with the histone H3K4 methylation complex at Pax target genes in renal cells. Thus, these compounds may provide structural scaffolds for kidney-specific inhibitors with therapeutic potential.
Collapse
Affiliation(s)
- Shayna T J Bradford
- Department of Pathology, University of Michigan, BSRB 2049, 109 Zina Pitcher Drive, Ann Arbor, MI 48109, USA; Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Edward Grimley
- Department of Pathology, University of Michigan, BSRB 2049, 109 Zina Pitcher Drive, Ann Arbor, MI 48109, USA; Molecular and Cellular Pathology Graduate Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ann M Laszczyk
- Department of Pathology, University of Michigan, BSRB 2049, 109 Zina Pitcher Drive, Ann Arbor, MI 48109, USA
| | - Pil H Lee
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sanjeevkumar R Patel
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Gregory R Dressler
- Department of Pathology, University of Michigan, BSRB 2049, 109 Zina Pitcher Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
14
|
Okamura DM, Brewer CM, Wakenight P, Bahrami N, Bernardi K, Tran A, Olson J, Shi X, Yeh SY, Piliponsky A, Collins SJ, Nguyen ED, Timms AE, MacDonald JW, Bammler TK, Nelson BR, Millen KJ, Beier DR, Majesky MW. Spiny mice activate unique transcriptional programs after severe kidney injury regenerating organ function without fibrosis. iScience 2021; 24:103269. [PMID: 34849462 PMCID: PMC8609232 DOI: 10.1016/j.isci.2021.103269] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 09/02/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Fibrosis-driven solid organ failure is an enormous burden on global health. Spiny mice (Acomys) are terrestrial mammals that can regenerate severe skin wounds without scars to avoid predation. Whether spiny mice also regenerate internal organ injuries is unknown. Here, we show that despite equivalent acute obstructive or ischemic kidney injury, spiny mice fully regenerate nephron structure and organ function without fibrosis, whereas C57Bl/6 or CD1 mice progress to complete organ failure with extensive renal fibrosis. Two mechanisms for vertebrate regeneration have been proposed that emphasize either extrinsic (pro-regenerative macrophages) or intrinsic (surviving cells of the organ itself) controls. Comparative transcriptome analysis revealed that the Acomys genome appears poised at the time of injury to initiate regeneration by surviving kidney cells, whereas macrophage accumulation was not detected until about day 7. Thus, we provide evidence for rapid activation of a gene expression signature for regenerative wound healing in the spiny mouse kidney.
Collapse
Affiliation(s)
- Daryl M. Okamura
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Chris M. Brewer
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Paul Wakenight
- Center for Integrated Brain Research, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Nadia Bahrami
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Kristina Bernardi
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Amy Tran
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Jill Olson
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Xiaogang Shi
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Szu-Ying Yeh
- Center for Integrated Brain Research, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Adrian Piliponsky
- Center for Immunity & Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Sarah J. Collins
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Elizabeth D. Nguyen
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Andrew E. Timms
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - James W. MacDonald
- Department of Environmental & Occupational Health, University of Washington, Seattle, WA 98195, USA
| | - Theo K. Bammler
- Department of Environmental & Occupational Health, University of Washington, Seattle, WA 98195, USA
| | - Branden R. Nelson
- Center for Integrated Brain Research, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Kathleen J. Millen
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Integrated Brain Research, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - David R. Beier
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
| | - Mark W. Majesky
- Department of Pediatrics, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98105, USA
- Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
15
|
Zhao Z, Li S, Li S, Wang J, Lin H, Fu W. High expression of oncogene cadherin-6 correlates with tumor progression and a poor prognosis in gastric cancer. Cancer Cell Int 2021; 21:493. [PMID: 34530820 PMCID: PMC8447617 DOI: 10.1186/s12935-021-02071-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most common and fatal cancers worldwide. Effective biomarkers to aid the early diagnosis of GC, as well as predict the course of disease, are urgently needed. Hence, we explored the role and function of cadherin-6 (CDH6) in the diagnosis and prognosis of gastric cancer. Methods The expression levels of CDH6 in cancerous and normal gastric tissue were analyzed using multiple public databases. Gene set enrichment analysis (GSEA) was performed using The Cancer Genome Atlas (TCGA) dataset. The diagnostic efficiency of CDH6 expression in GC patients was determined through receiver operating characteristic (ROC) curve analysis. The associations between clinical variables and CDH6 expression were evaluated statistically, and the prognostic factors for overall survival were analyzed by univariate and multivariate Cox regression. 44 GC tissue samples, 20 donor-matched adjacent normal tissue samples, and associated detailed clinical information, were collected from the Tianjin Medical University General Hospital. CDH6 expression levels were determined for further validation. Results CDH6 was upregulated in GC samples compared to normal gastric tissue. Furthermore, GSEA identified the tricarboxylic acid (TCA) cycle, extracellular matrix (ECM) receptor interaction, glyoxylate and dicarboxylate metabolism, oxidative phosphorylation, and the pentose phosphate pathway as differentially enriched in GC tissue samples. According to the area under the ROC curve (AUC) values (AUC = 0.829 in the TCGA and 0.966 in the GSE54129 dataset), CDH6 expression was associated with high diagnostic efficacy. Patients with high CDH6 levels in their GC tissues had a higher T number (according to the TNM classification) and a worse prognosis than those with low CDH6 expression. Univariate and multivariate Cox regression analysis showed that CDH6 was an independent risk factor for overall survival (univariate: HR = 1.305, P = 0.002, multivariate: HR = 1.481, P < 0.001). Conclusion CDH6 was upregulated in GC, and high CDH6 expression was indicative of a higher T number and a worse prognosis. Therefore, CDH6 represents a potentially independent molecular biomarker for the diagnostic and prognostic prediction of GC. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02071-y.
Collapse
Affiliation(s)
- Zongxian Zhao
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China.
| | - Shuliang Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Liaocheng, Liaocheng, Shangdong, China.
| | - Shilong Li
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China
| | - Jun Wang
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China
| | - Hai Lin
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China
| | - Weihua Fu
- Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, China.
| |
Collapse
|
16
|
Lindström NO, Sealfon R, Chen X, Parvez RK, Ransick A, De Sena Brandine G, Guo J, Hill B, Tran T, Kim AD, Zhou J, Tadych A, Watters A, Wong A, Lovero E, Grubbs BH, Thornton ME, McMahon JA, Smith AD, Ruffins SW, Armit C, Troyanskaya OG, McMahon AP. Spatial transcriptional mapping of the human nephrogenic program. Dev Cell 2021; 56:2381-2398.e6. [PMID: 34428401 PMCID: PMC8396064 DOI: 10.1016/j.devcel.2021.07.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 05/06/2021] [Accepted: 07/27/2021] [Indexed: 12/11/2022]
Abstract
Congenital abnormalities of the kidney and urinary tract are among the most common birth defects, affecting 3% of newborns. The human kidney forms around a million nephrons from a pool of nephron progenitors over a 30-week period of development. To establish a framework for human nephrogenesis, we spatially resolved a stereotypical process by which equipotent nephron progenitors generate a nephron anlage, then applied data-driven approaches to construct three-dimensional protein maps on anatomical models of the nephrogenic program. Single-cell RNA sequencing identified progenitor states, which were spatially mapped to the nephron anatomy, enabling the generation of functional gene networks predicting interactions within and between nephron cell types. Network mining identified known developmental disease genes and predicted targets of interest. The spatially resolved nephrogenic program made available through the Human Nephrogenesis Atlas (https://sckidney.flatironinstitute.org/) will facilitate an understanding of kidney development and disease and enhance efforts to generate new kidney structures.
Collapse
Affiliation(s)
- Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Rachel Sealfon
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Xi Chen
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew Ransick
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guilherme De Sena Brandine
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern, Los Angeles, CA 90089, USA
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bill Hill
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jian Zhou
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Alicja Tadych
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Aaron Watters
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Aaron Wong
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Elizabeth Lovero
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA
| | - Brendan H Grubbs
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andrew D Smith
- Molecular and Computational Biology, Division of Biological Sciences, University of Southern, Los Angeles, CA 90089, USA
| | - Seth W Ruffins
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Chris Armit
- MRC Human Genetics Unit, MRC Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK; BGI Hong Kong, 26/F, Kings Wing Plaza 2, 1 On Kwan Street, Shek Mun, NT, Hong Kong
| | - Olga G Troyanskaya
- Center for Computational Biology, Flatiron Institute, Simons Foundation, New York, NY, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA; Department of Computer Science, Princeton University, Princeton, NJ, USA.
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Ide S, Kobayashi Y, Ide K, Strausser SA, Abe K, Herbek S, O'Brien LL, Crowley SD, Barisoni L, Tata A, Tata PR, Souma T. Ferroptotic stress promotes the accumulation of pro-inflammatory proximal tubular cells in maladaptive renal repair. eLife 2021; 10:68603. [PMID: 34279220 PMCID: PMC8318592 DOI: 10.7554/elife.68603] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 07/17/2021] [Indexed: 12/14/2022] Open
Abstract
Overwhelming lipid peroxidation induces ferroptotic stress and ferroptosis, a non-apoptotic form of regulated cell death that has been implicated in maladaptive renal repair in mice and humans. Using single-cell transcriptomic and mouse genetic approaches, we show that proximal tubular (PT) cells develop a molecularly distinct, pro-inflammatory state following injury. While these inflammatory PT cells transiently appear after mild injury and return to their original state without inducing fibrosis, after severe injury they accumulate and contribute to persistent inflammation. This transient inflammatory PT state significantly downregulates glutathione metabolism genes, making the cells vulnerable to ferroptotic stress. Genetic induction of high ferroptotic stress in these cells after mild injury leads to the accumulation of the inflammatory PT cells, enhancing inflammation and fibrosis. Our study broadens the roles of ferroptotic stress from being a trigger of regulated cell death to include the promotion and accumulation of proinflammatory cells that underlie maladaptive repair.
Collapse
Affiliation(s)
- Shintaro Ide
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Yoshihiko Kobayashi
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Kana Ide
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Sarah A Strausser
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Koki Abe
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Savannah Herbek
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Lori L O'Brien
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Steven D Crowley
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States
| | - Laura Barisoni
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States.,Department of Pathology, Duke University School of Medicine, Durham, United States
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, United States
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, United States.,Regeneration Next, Duke University, Durham, United States.,Duke Cancer Institute, Duke University School of Medicine, Durham, United States
| | - Tomokazu Souma
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, United States.,Regeneration Next, Duke University, Durham, United States
| |
Collapse
|
18
|
Cornelissen A, Guo L, Fernandez R, Kelly MC, Janifer C, Kuntz S, Sakamoto A, Jinnouchi H, Sato Y, Paek KH, Kolodgie FD, Romero ME, Surve D, Virmani R, Finn AV. Endothelial Recovery in Bare Metal Stents and Drug-Eluting Stents on a Single-Cell Level. Arterioscler Thromb Vasc Biol 2021; 41:2277-2292. [PMID: 34162228 DOI: 10.1161/atvbaha.121.316472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anne Cornelissen
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,Department of Cardiology, University Hospital RWTH Aachen, Germany (A.C.)
| | - Liang Guo
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Raquel Fernandez
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Michael C Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD (M.C.K.)
| | - Christine Janifer
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Salome Kuntz
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Atsushi Sakamoto
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Hiroyuki Jinnouchi
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Yu Sato
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Ka Hyun Paek
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Frank D Kolodgie
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Maria E Romero
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Dipti Surve
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Renu Virmani
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,University of Maryland, School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
19
|
Chandrasekaran V, Carta G, da Costa Pereira D, Gupta R, Murphy C, Feifel E, Kern G, Lechner J, Cavallo AL, Gupta S, Caiment F, Kleinjans JCS, Gstraunthaler G, Jennings P, Wilmes A. Generation and characterization of iPSC-derived renal proximal tubule-like cells with extended stability. Sci Rep 2021; 11:11575. [PMID: 34078926 PMCID: PMC8172841 DOI: 10.1038/s41598-021-89550-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 04/23/2021] [Indexed: 12/21/2022] Open
Abstract
The renal proximal tubule is responsible for re-absorption of the majority of the glomerular filtrate and its proper function is necessary for whole-body homeostasis. Aging, certain diseases and chemical-induced toxicity are factors that contribute to proximal tubule injury and chronic kidney disease progression. To better understand these processes, it would be advantageous to generate renal tissues from human induced pluripotent stem cells (iPSC). Here, we report the differentiation and characterization of iPSC lines into proximal tubular-like cells (PTL). The protocol is a step wise exposure of small molecules and growth factors, including the GSK3 inhibitor (CHIR99021), the retinoic acid receptor activator (TTNPB), FGF9 and EGF, to drive iPSC to PTL via cell stages representing characteristics of early stages of renal development. Genome-wide RNA sequencing showed that PTL clustered within a kidney phenotype. PTL expressed proximal tubular-specific markers, including megalin (LRP2), showed a polarized phenotype, and were responsive to parathyroid hormone. PTL could take up albumin and exhibited ABCB1 transport activity. The phenotype was stable for up to 7 days and was maintained after passaging. This protocol will form the basis of an optimized strategy for molecular investigations using iPSC derived PTL.
Collapse
Affiliation(s)
- Vidya Chandrasekaran
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Giada Carta
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Daniel da Costa Pereira
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Rajinder Gupta
- Department of Toxicogenomics, Maastricht University, School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Cormac Murphy
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands
| | - Elisabeth Feifel
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Georg Kern
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Judith Lechner
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | | | | | - Florian Caiment
- Department of Toxicogenomics, Maastricht University, School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Jos C S Kleinjans
- Department of Toxicogenomics, Maastricht University, School of Oncology and Developmental Biology (GROW), Maastricht, The Netherlands
| | - Gerhard Gstraunthaler
- Institute of Physiology and Medical Physics, Medical University of Innsbruck, Innsbruck, Austria
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands.
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081, HZ, Amsterdam, The Netherlands.
| |
Collapse
|
20
|
Thomson RB, Dynia DW, Burlein S, Thomson BR, Booth CJ, Knauf F, Wang T, Aronson PS. Deletion of Cdh16 Ksp-cadherin leads to a developmental delay in the ability to maximally concentrate urine in mouse. Am J Physiol Renal Physiol 2021; 320:F1106-F1122. [PMID: 33938239 DOI: 10.1152/ajprenal.00556.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ksp-cadherin (cadherin-16) is an atypical member of the cadherin superfamily of cell adhesion molecules that is ubiquitously expressed on the basolateral membrane of epithelial cells lining the nephron and the collecting system of the mammalian kidney. The principal aim of the present study was to determine if Ksp-cadherin played a critical role in the development and maintenance of the adult mammalian kidney by generating and evaluating a mouse line deficient in Ksp-cadherin. Ksp-null mutant animals were viable and fertile, and kidneys from both neonates and adults showed no evidence of structural abnormalities. Immunolocalization and Western blot analyses of Na+-K+-ATPase and E-cadherin indicated that Ksp-cadherin is not essential for either the genesis or maintenance of the polarized tubular epithelial phenotype. Moreover, E-cadherin expression was not altered to compensate for Ksp-cadherin loss. Plasma electrolytes, total CO2, blood urea nitrogen, and creatinine levels were also unaffected by Ksp-cadherin deficiency. However, a subtle but significant developmental delay in the ability to maximally concentrate urine was detected in Ksp-null mice. Expression analysis of the principal proteins involved in the generation of the corticomedullary osmotic gradient and the resultant movement of water identified misexpression of aquaporin-2 in the inner medullary collecting duct as the possible cause for the inability of young adult Ksp-cadherin-deficient animals to maximally concentrate their urine. In conclusion, Ksp-cadherin is not required for normal kidney development, but its absence leads to a developmental delay in maximal urinary concentrating ability.NEW & NOTEWORTHY Ksp-cadherin (cadherin-16) is an atypical member of the cadherin superfamily of cell adhesion molecules that is ubiquitously expressed on the basolateral membrane of epithelial cells lining the nephron and the collecting system. Using knockout mice, we found that Ksp-cadherin is in fact not required for kidney development despite its high and specific expression along the nephron. However, its absence leads to a developmental delay in maximal urinary concentrating ability.
Collapse
Affiliation(s)
- R B Thomson
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - D W Dynia
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - S Burlein
- Department of Nephrology and Hypertension, Friedrich Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - B R Thomson
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - C J Booth
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - F Knauf
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - T Wang
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| | - P S Aronson
- Department of Internal Medicine, Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
21
|
Chambers JM, Wingert RA. Advances in understanding vertebrate nephrogenesis. Tissue Barriers 2020; 8:1832844. [PMID: 33092489 PMCID: PMC7714473 DOI: 10.1080/21688370.2020.1832844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/01/2020] [Accepted: 10/01/2020] [Indexed: 02/07/2023] Open
Abstract
The kidney is a complex organ that performs essential functions such as blood filtration and fluid homeostasis, among others. Recent years have heralded significant advancements in our knowledge of the mechanisms that control kidney formation. Here, we provide an overview of vertebrate renal development with a focus on nephrogenesis, the process of generating the epithelialized functional units of the kidney. These steps begin with intermediate mesoderm specification and proceed all the way to the terminally differentiated nephron cell, with many detailed stages in between. The establishment of nephron architecture with proper cellular barriers is vital throughout these processes. Continuously striving to gain further insights into nephrogenesis can ultimately lead to a better understanding and potential treatments for developmental maladies such as Congenital Anomalies of the Kidney and Urinary Tract (CAKUT).
Collapse
Affiliation(s)
- Joseph M. Chambers
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| | - Rebecca A. Wingert
- Department of Biological Sciences, Center for Stem Cells and Regenerative Medicine, Center for Zebrafish Research, Boler-Parseghian Center for Rare and Neglected Diseases, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
22
|
Beyond N-Cadherin, Relevance of Cadherins 5, 6 and 17 in Cancer Progression and Metastasis. Int J Mol Sci 2019; 20:ijms20133373. [PMID: 31324051 PMCID: PMC6651558 DOI: 10.3390/ijms20133373] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/01/2019] [Accepted: 07/06/2019] [Indexed: 12/12/2022] Open
Abstract
Cell-cell adhesion molecules (cadherins) and cell-extracellular matrix adhesion proteins (integrins) play a critical role in the regulation of cancer invasion and metastasis. Although significant progress has been made in the characterization of multiple members of the cadherin superfamily, most of the published work continues to focus in the switch E-/N-cadherin and its role in the epithelial-mesenchymal transition. Here, we will discuss the structural and functional properties of a subset of cadherins (cadherin 17, cadherin 5 and cadherin 6) that have an RGD motif in the extracellular domains. This RGD motif is critical for the interaction with α2β1 integrin and posterior integrin pathway activation in cancer metastatic cells. However, other signaling pathways seem to be affected by RGD cadherin interactions, as will be discussed. The range of solid tumors with overexpression or "de novo" expression of one or more of these three cadherins is very wide (gastrointestinal, gynaecological and melanoma, among others), underscoring the relevance of these cadherins in cancer metastasis. Finally, we will discuss different evidences that support the therapeutic use of these cadherins by blocking their capacity to work as integrin ligands in order to develop new cures for metastatic patients.
Collapse
|
23
|
Yang YK, Shen DD, He P, Du LD, Wan DJ, Wang P, Wang T, Feng MQ. Chemically synthesized LYRM03 could inhibit the metastasis of human breast cancer MDA-MB-231 cells in vitro and in vivo. Bioorg Med Chem Lett 2019; 29:1719-1726. [PMID: 31126854 DOI: 10.1016/j.bmcl.2019.05.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 05/15/2019] [Indexed: 02/02/2023]
Abstract
Aminopeptidase N (APN) belongs to the aminopeptidase family, which is widely distributed throughout the animal and plant kingdoms. APN is thought to be a very important target for cancer therapy as it is linked to cancer progression and metastasis. However, bestatin (Ubenimex) is the only approved drug that targets various aminopeptidases for the treatment of acute myelocytic leukemia and lymphedema. A compound 3-amino-2-hydroxy-4-phenylbutanoylvalylisoleucine (also known as LYRM03), isolated from a Streptomyces strain HCCB10043, exhibited more potent inhibitory activity than bestatin. In this work, we applied a chemical synthesis strategy to generate LYRM03 to overcome the low yields typically achieved from fermentation. Finally, we explored a suite of experiments to determine the bioactivity of LYRM03 and revealed that the metastasis of MDA-MB-231 cells was significantly restrained with LYRM03 treatment or injection both in vitro and in vivo. Because of its anti-metastasis capacity, further structure modifications of LYRM03 will be of interest for its use alone or in combination as a therapy in cancer.
Collapse
Affiliation(s)
- Yun-Kai Yang
- Department of Microbiology and Biochemical Pharmacy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Da-Dong Shen
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China
| | - Peng He
- Department of Microbiology and Biochemical Pharmacy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Liang-Dong Du
- Shanghai Laiyi Center for Biopharmaceuticals R&D, 5B, Building 8 200 Niudun Road Pudong District, Shanghai 201203, PR China
| | - Ding-Jian Wan
- Shanghai Laiyi Center for Biopharmaceuticals R&D, 5B, Building 8 200 Niudun Road Pudong District, Shanghai 201203, PR China
| | - Pu Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China
| | - Tao Wang
- Shanghai Laiyi Center for Biopharmaceuticals R&D, 5B, Building 8 200 Niudun Road Pudong District, Shanghai 201203, PR China.
| | - Mei-Qing Feng
- Department of Microbiology and Biochemical Pharmacy, School of Pharmacy, Fudan University, Shanghai 201203, PR China.
| |
Collapse
|
24
|
Ye J, He J, Li N. Molecular identification and characterization of pig's Cdh16 gene. GENE REPORTS 2019. [DOI: 10.1016/j.genrep.2018.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Harder JL, Menon R, Otto EA, Zhou J, Eddy S, Wys NL, O'Connor C, Luo J, Nair V, Cebrian C, Spence JR, Bitzer M, Troyanskaya OG, Hodgin JB, Wiggins RC, Freedman BS, Kretzler M. Organoid single cell profiling identifies a transcriptional signature of glomerular disease. JCI Insight 2019; 4:122697. [PMID: 30626756 PMCID: PMC6485369 DOI: 10.1172/jci.insight.122697] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/27/2018] [Indexed: 12/13/2022] Open
Abstract
Podocyte injury is central to many forms of kidney disease, but transcriptional signatures reflecting podocyte injury and compensation mechanisms are challenging to analyze in vivo. Human kidney organoids derived from pluripotent stem cells (PSCs), a potentially new model for disease and regeneration, present an opportunity to explore the transcriptional plasticity of podocytes. Here, transcriptional profiling of more than 12,000 single cells from human PSC-derived kidney organoid cultures was used to identify robust and reproducible cell lineage gene expression signatures shared with developing human kidneys based on trajectory analysis. Surprisingly, the gene expression signature characteristic of developing glomerular epithelial cells was also observed in glomerular tissue from a kidney disease cohort. This signature correlated with proteinuria and inverse eGFR, and it was confirmed in an independent podocytopathy cohort. Three genes in particular were further characterized as potentially novel components of the glomerular disease signature. We conclude that cells in human PSC-derived kidney organoids reliably recapitulate the developmental transcriptional program of podocytes and other cell lineages in the human kidney and that transcriptional profiles seen in developing podocytes are reactivated in glomerular disease. Our findings demonstrate an approach to identifying potentially novel molecular programs involved in the pathogenesis of glomerulopathies.
Collapse
Affiliation(s)
| | - Rajasree Menon
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Edgar A Otto
- Department of Internal Medicine, Division of Nephrology, and
| | - Jian Zhou
- Flatiron Institute, Simons Foundation, New York, New York, USA
| | - Sean Eddy
- Department of Internal Medicine, Division of Nephrology, and
| | - Noel L Wys
- Department of Internal Medicine, Division of Nephrology, and
| | | | | | - Viji Nair
- Department of Internal Medicine, Division of Nephrology, and
| | - Cristina Cebrian
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R Spence
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Markus Bitzer
- Department of Internal Medicine, Division of Nephrology, and
| | - Olga G Troyanskaya
- Flatiron Institute, Simons Foundation, New York, New York, USA.,Lewis-Sigler Institute for Integrative Genomics and.,Department of Computer Science, Princeton University, Princeton, New Jersey, USA
| | | | - Roger C Wiggins
- Department of Internal Medicine, Division of Nephrology, and
| | - Benjamin S Freedman
- Department of Medicine, Division of Nephrology.,Kidney Research Institute.,Institute for Stem Cell and Regenerative Medicine, and.,Department of Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthias Kretzler
- Department of Internal Medicine, Division of Nephrology, and.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
26
|
Duan X, Krishnaswamy A, Laboulaye MA, Liu J, Peng YR, Yamagata M, Toma K, Sanes JR. Cadherin Combinations Recruit Dendrites of Distinct Retinal Neurons to a Shared Interneuronal Scaffold. Neuron 2018; 99:1145-1154.e6. [PMID: 30197236 PMCID: PMC6284407 DOI: 10.1016/j.neuron.2018.08.019] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022]
Abstract
Distinct neuronal types connect in complex ways to generate functional neural circuits. The molecular diversity required to specify this connectivity could be supplied by multigene families of synaptic recognition molecules, but most studies to date have assessed just one or a few members at a time. Here, we analyze roles of cadherins (Cdhs) in formation of retinal circuits comprising eight neuronal types that inform the brain about motion in four directions. We show that at least 15 classical Cdhs are expressed by neurons in these circuits and at least 6 (Cdh6-10 and 18) act individually or in combinations to promote specific connectivity among the cells. They act in part by directing the processes of output neurons and excitatory interneurons to a cellular scaffold formed by inhibitory interneurons. Because Cdhs are expressed combinatorially by many central neurons, similar interactions could be involved in patterning circuits throughout the brain.
Collapse
Affiliation(s)
- Xin Duan
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA; Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Arjun Krishnaswamy
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Mallory A Laboulaye
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jinyue Liu
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Yi-Rong Peng
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Masahito Yamagata
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Kenichi Toma
- Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
27
|
Gong X, Guo X, Huang R, Liao H, Zhang Q, Yan J, Luo L, Zhang Q, Qiu A, Sun Y, Liang X. Expression of ILK in renal stroma is essential for multiple aspects of renal development. Am J Physiol Renal Physiol 2018; 315:F374-F385. [PMID: 29638158 DOI: 10.1152/ajprenal.00509.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Kidney development involves reciprocal and inductive interactions between the ureteric bud (UB) and surrounding metanephric mesenchyme. Signals from renal stromal lineages are essential for differentiation and patterning of renal epithelial and mesenchymal cell types and renal vasculogenesis; however, underlying mechanisms remain not fully understood. Integrin-linked kinase (ILK), a key component of integrin signaling pathway, plays an important role in kidney development. However, the role of ILK in renal stroma remains unknown. Here, we ablated ILK in renal stromal lineages using a platelet-derived growth factor receptor B ( Pdgfrb) -Cre mouse line, and the resulting Ilk mutant mice presented postnatal growth retardation and died within 3 wk of age with severe renal developmental defects. Pdgfrb-Cre;Ilk mutant kidneys exhibited a significant decrease in UB branching and disrupted collecting duct formation. From E16.5 onward, renal interstitium was disorganized, forming medullary interstitial pseudocysts. Pdgfrb-Cre;Ilk mutants exhibited renal vasculature mispatterning and impaired glomerular vascular differentiation. Impaired glial cell-derived neurotrophic factor/Ret and bone morphogenetic protein 7 signaling pathways were observed in Pdgfrb-Cre;Ilk mutant kidneys. Furthermore, phosphoproteomic and Western blot analyses revealed a significant dysregulation of a number of key signaling pathways required for kidney morphogenesis, including PI3K/AKT and MAPK/ERK in Pdgfrb-Cre;Ilk mutants. Our results revealed a critical requirement for ILK in renal-stromal and vascular development, as well as a noncell autonomous role of ILK in UB branching morphogenesis.
Collapse
Affiliation(s)
- Xiaohui Gong
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Xiaoxia Guo
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Ru Huang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Huimin Liao
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Qingquan Zhang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Jie Yan
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Lina Luo
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Qitong Zhang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Andong Qiu
- School of Life Sciences and Technology, Tongji University , Shanghai , China
| | - Yunfu Sun
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| | - Xingqun Liang
- Key Laboratory of Arrhythmia, Ministry of Education, East Hospital, School of Medicine, Tongji University , Shanghai , China
| |
Collapse
|
28
|
Przepiorski A, Sander V, Tran T, Hollywood JA, Sorrenson B, Shih JH, Wolvetang EJ, McMahon AP, Holm TM, Davidson AJ. A Simple Bioreactor-Based Method to Generate Kidney Organoids from Pluripotent Stem Cells. Stem Cell Reports 2018; 11:470-484. [PMID: 30033089 PMCID: PMC6092837 DOI: 10.1016/j.stemcr.2018.06.018] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 02/07/2023] Open
Abstract
Kidney organoids made from pluripotent stem cells have the potential to revolutionize how kidney development, disease, and injury are studied. Current protocols are technically complex, suffer from poor reproducibility, and have high reagent costs that restrict scalability. To overcome some of these issues, we have established a simple, inexpensive, and robust method to grow kidney organoids in bulk from human induced pluripotent stem cells. Our organoids develop tubular structures by day 8 and show optimal tissue morphology at day 14. A comparison with fetal human kidneys suggests that day-14 organoid tissue most closely resembles late capillary loop stage nephrons. We show that deletion of HNF1B, a transcription factor linked to congenital kidney defects, interferes with tubulogenesis, validating our experimental system for studying renal developmental biology. Taken together, our protocol provides a fast, efficient, and cost-effective method for generating large quantities of human fetal kidney tissue, enabling the study of normal and aberrant kidney development. Technically simple and cost-efficient protocol for kidney organoid generation Tubular organoids are obtained rapidly, with high efficiency, yield, and robustness Organoids contain nephrons that correspond to human fetal nephrons The applicability to model congenital kidney defects is presented
Collapse
Affiliation(s)
- Aneta Przepiorski
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand
| | - Veronika Sander
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Jennifer A Hollywood
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand
| | - Brie Sorrenson
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand
| | - Jen-Hsing Shih
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD 4072, Australia
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Teresa M Holm
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand
| | - Alan J Davidson
- Department of Molecular Medicine & Pathology, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
29
|
Geometry of Gene Expression Space of Wilms' Tumors From Human Patients. Neoplasia 2018; 20:871-881. [PMID: 30029183 PMCID: PMC6076422 DOI: 10.1016/j.neo.2018.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 02/05/2023] Open
Abstract
Wilms' tumor is a pediatric malignancy that is thought to originate from faulty kidney development during the embryonic stage. However, there is a large variation between tumors from different patients in both histology and gene expression that is not well characterized. Here we use a meta-analysis of published microarray datasets to show that Favorable Histology Wilms' Tumors (FHWT's) fill a triangle-shaped continuum in gene expression space of which the vertices represent three idealized “archetypes”. We show that these archetypes have predominantly renal blastemal, stromal, and epithelial characteristics and that they correlate well with the three major lineages of the developing embryonic kidney. Moreover, we show that advanced stage tumors shift towards the renal blastemal archetype. These results illustrate the potential of this methodology for characterizing the cellular composition of Wilms' tumors and for assessing disease progression.
Collapse
|
30
|
Casal JI, Bartolomé RA. RGD cadherins and α2β1 integrin in cancer metastasis: A dangerous liaison. Biochim Biophys Acta Rev Cancer 2018; 1869:321-332. [PMID: 29673969 DOI: 10.1016/j.bbcan.2018.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/24/2022]
Abstract
We propose a new cadherin family classification comprising epithelial cadherins (cadherin 17 [CDH17], cadherin 16, VE-cadherin, cadherin 6 and cadherin 20) containing RGD motifs within their sequences. Expression of some RGD cadherins is associated with aggressive forms of cancer during the late stages of metastasis, and CDH17 and VE-cadherin have emerged as critical actors in cancer metastasis. After binding to α2β1 integrin, these cadherins promote integrin β1 activation, and thereby cell adhesion, invasion and proliferation, in liver and lung metastasis. Activation of α2β1 integrin provokes an affinity increase for type IV collagen, a major component of the basement membrane and a critical partner for cell anchoring in liver and other metastatic organs. Activation of α2β1 integrin by RGD motifs breaks an old paradigm of integrin classification and supports an important role of this integrin in cancer metastasis. Recently, synthetic peptides containing the RGD motif of CDH17 elicited highly specific and selective antibodies that block the ability of CDH17 RGD to activate α2β1 integrin. These monoclonal antibodies inhibit metastatic colonization in orthotopic mouse models of liver and lung metastasis for colorectal cancer and melanoma, respectively. Hopefully, blocking the cadherin RGD ligand capacity will give us control over the integrin activity in solid tumors metastasis, paving the way for development of new agents of cancer treatment.
Collapse
Affiliation(s)
- J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039 Madrid, Spain.
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039 Madrid, Spain
| |
Collapse
|
31
|
Lindström NO, Tran T, Guo J, Rutledge E, Parvez RK, Thornton ME, Grubbs B, McMahon JA, McMahon AP. Conserved and Divergent Molecular and Anatomic Features of Human and Mouse Nephron Patterning. J Am Soc Nephrol 2018; 29:825-840. [PMID: 29449451 PMCID: PMC5827611 DOI: 10.1681/asn.2017091036] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/27/2017] [Indexed: 11/03/2022] Open
Abstract
The nephron is the functional unit of the kidney, but the mechanism of nephron formation during human development is unclear. We conducted a detailed analysis of nephron development in humans and mice by immunolabeling, and we compared human and mouse nephron patterning to describe conserved and divergent features. We created protein localization maps that highlight the emerging patterns along the proximal-distal axis of the developing nephron and benchmark expectations for localization of functionally important transcription factors, which revealed unanticipated cellular diversity. Moreover, we identified a novel nephron subdomain marked by Wnt4 expression that we fate-mapped to the proximal mature nephron. Significant conservation was observed between human and mouse patterning. We also determined the time at which markers for mature nephron cell types first emerge-critical data for the renal organoid field. These findings have conceptual implications for the evolutionary processes driving the diversity of mammalian organ systems. Furthermore, these findings provide practical insights beyond those gained with mouse and rat models that will guide in vitro efforts to harness the developmental programs necessary to build human kidney structures.
Collapse
Affiliation(s)
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine and
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine and
| | | | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine and
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine and
| | | |
Collapse
|
32
|
Cadherin 6 is activated by Epstein-Barr virus LMP1 to mediate EMT and metastasis as an interplay node of multiple pathways in nasopharyngeal carcinoma. Oncogenesis 2017; 6:402. [PMID: 29284791 PMCID: PMC5865538 DOI: 10.1038/s41389-017-0005-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/23/2017] [Accepted: 10/01/2017] [Indexed: 12/14/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy, which is notorious among head-and-neck cancers with its metastatic feature. Epstein–Barr virus (EBV) infection plays a fundamental role in NPC development with the mechanism is not well understood. Here we demonstrate that EBV oncoprotein LMP1 drives EMT and metastasis of NPC by reactivating the adhesion molecule, cadherin 6 (CDH6), which normally occurs in embryogenesis with unknown role in NPC. CDH6 was found to be upregulated in LMP1-positive NPC tissues, and was identified as a target of the epithelium-specific miR-203. LMP1-activated NF-κB transcriptionally repressed the miR-203 expression by binding to the promoter region of miR-203 gene. CDH6 activation in turn induced EMT and promoted metastasis in NPC. CDH6 depletion, NF-κB inhibitor and miR-203 overexpression were able to impair the EMT effects. The miR-203 downregulation in NPC tissues was strongly associated with metastasis clinically. The CDH6 activator, Runt-related transcription factor 2 (RUNX2), was also activated by EBV in the event. For both CDH6 and RUNX2 are components at TGF-β downstream, CDH6 became a node protein for the interplay of multiple signalings including NF-κB and TGF-β. Therefore, the switch-on of miR-203 was important for nasopharyngeal epithelial cells to maintain normal phenotype. This study demonstrates that EBV has evolved sophisticated strategies by driving epithelial cells to obtain malignant features, particularly in NPC metastasis, providing novel biomarkers for the therapy and prognosis of EBV-associated NPC.
Collapse
|
33
|
Chung E, Deacon P, Park JS. Notch is required for the formation of all nephron segments and primes nephron progenitors for differentiation. Development 2017; 144:4530-4539. [PMID: 29113990 DOI: 10.1242/dev.156661] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/26/2017] [Indexed: 12/14/2022]
Abstract
Notch signaling plays important roles during mammalian nephrogenesis. To investigate whether Notch regulates nephron segmentation, we performed Notch loss-of-function and gain-of-function studies in developing nephrons in mice. Contrary to the previous notion that Notch signaling promotes the formation of proximal tubules and represses the formation of distal tubules in the mammalian nephron, we show that inhibition of Notch blocks the formation of all nephron segments and that constitutive activation of Notch in developing nephrons does not promote or repress the formation of a specific segment. Cells lacking Notch fail to form the S-shaped body and show reduced expression of Lhx1 and Hnf1b Consistent with this, we find that constitutive activation of Notch in mesenchymal nephron progenitors causes ectopic expression of Lhx1 and Hnf1b and that these cells eventually form a heterogeneous population that includes proximal tubules and other types of cells. Our data suggest that Notch signaling is required for the formation of all nephron segments and that it primes nephron progenitors for differentiation rather than directing their cell fates into a specific nephron segment.
Collapse
Affiliation(s)
- Eunah Chung
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Patrick Deacon
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Joo-Seop Park
- Division of Pediatric Urology and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
34
|
Terada N, Karim MR, Izawa T, Kuwamura M, Yamate J. Immunolocalization of β-catenin, E-cadherin and N-cadherin in neonate and adult rat kidney. J Vet Med Sci 2017; 79:1785-1790. [PMID: 28993569 PMCID: PMC5709553 DOI: 10.1292/jvms.17-0439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
β-catenin, E-cadherin and N-cadherin are adhesion molecules that play important roles in organogenesis, tissue homeostasis, renal epithelial integrity and polarity. The present study demonstrated their immunolocalization in adult and neonate rat kidney. Membranous or cytoplasmic expression of β-catenin, E-cadherin and N-cadherin were seen in adult and developing renal tubular epithelial cells. Particularly, in adult kidney, E-cadherin and β-catenin were intensively expressed in distal renal tubules, whereas N-cadherin was expressed in proximal renal tubules. In neonate rat kidney on 1 and 4 days old, developing renal tubular epithelial cells were mainly reacted with E-cadherin and very weakly expressed N-cadherin; β-catenin was expressed in developing renal tubules and mesenchymal blastemal cells. Interestingly, β-catenin-positive renal tubular epithelial cells simultaneously expressed E-cadherin in the kidney of adult and developing rats. Collectively, the adhesion molecules were differentially distributed in the renal tubules of adult rats and β-catenin and E-cadherin are predominant adhesion molecules in developing kidney. The present findings would provide the basic information of evaluating renal tubular toxicity using rats, in addition to renal genesis, in terms of adhesion molecules.
Collapse
Affiliation(s)
- Naomi Terada
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano-shi, Osaka 598-8531, Japan
| | - Mohammad Rabiul Karim
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano-shi, Osaka 598-8531, Japan
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano-shi, Osaka 598-8531, Japan
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano-shi, Osaka 598-8531, Japan
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-Ourai-Kita, Izumisano-shi, Osaka 598-8531, Japan
| |
Collapse
|
35
|
Tarrant KJ, Dey S, Kinney R, Anthony NB, Rhoads DD. Multi-generational genome wide association studies identify chromosomal regions associated with ascites phenotype. Poult Sci 2017; 96:1544-1552. [PMID: 28339749 PMCID: PMC5850653 DOI: 10.3382/ps/pew478] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 12/23/2016] [Indexed: 12/29/2022] Open
Abstract
Ascites is a multi-faceted disease commonly observed in fast growing broilers, which is initiated when the body is insufficiently oxygenated. A series of events follow, including an increase in pulmonary artery pressure, right ventricle hypertrophy, and accumulation of fluid in the abdominal cavity and pericardium. Advances in management practices along with improved selection programs have decreased ascites incidence in modern broilers. However, ascites syndrome remains an economically important disease throughout the world, causing estimated losses of $100 million per year. In this study, a 60 K Illumina SNP BeadChip was used to perform a series of genome wide association studies (GWAS) on the 16th and 18th generation of our relaxed (REL) line descended from a commercial elite broiler line beginning in 1995. Regions significantly associated with ascites incidence were identified on chromosome 2 around 70 megabase pairs (Mbp) and on chromosome Z around 60 Mbp. Five candidate single nucleotide polymorphisms (SNP) were evaluated as indicators for these 2 regions in order to identify association with ascites and right ventricle to total ventricle weight (RVTV) ratios. Chromosome 2 SNP showed an association with RVTV ratios in males phenotyped as ascites resistant and ascites susceptible (P = 0.02 and P = 0.03, respectively). The chromosome Z region also indicates an association with resistant female RVTV values (P = 0.02). Regions of significance identified on chromosomes 2 and Z described in this study will be used as proposed candidate regions for further investigation into the genetics of ascites. This information will lead to a better understanding of the underlying genetics and gene networks contributing to ascites, and thus advances in ascites reduction through commercial breeding schemes.
Collapse
Affiliation(s)
- K. J. Tarrant
- Department of Animal Sciences and Agricultural Education, California State University Fresno, Fresno 93740
| | - S. Dey
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville 72701
| | - R. Kinney
- John Brown University, Siloam Springs, AR 72761
| | - N. B. Anthony
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville 72701
- Department of Poultry Sciences, University of Arkansas, Fayetteville 72701
| | - D. D. Rhoads
- Program in Cell and Molecular Biology, University of Arkansas, Fayetteville 72701
- Department of Biological Sciences, University of Arkansas, Fayetteville 72701
| |
Collapse
|
36
|
Pode-Shakked N, Gershon R, Tam G, Omer D, Gnatek Y, Kanter I, Oriel S, Katz G, Harari-Steinberg O, Kalisky T, Dekel B. Evidence of In Vitro Preservation of Human Nephrogenesis at the Single-Cell Level. Stem Cell Reports 2017; 9:279-291. [PMID: 28552604 PMCID: PMC5511042 DOI: 10.1016/j.stemcr.2017.04.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 04/24/2017] [Accepted: 04/25/2017] [Indexed: 01/09/2023] Open
Abstract
During nephrogenesis, stem/progenitor cells differentiate and give rise to early nephron structures that segment to proximal and distal nephron cell types. Previously, we prospectively isolated progenitors from human fetal kidney (hFK) utilizing a combination of surface markers. However, upon culture nephron progenitors differentiated and could not be robustly maintained in vitro. Here, by culturing hFK in a modified medium used for in vitro growth of mouse nephron progenitors, and by dissection of NCAM+/CD133− progenitor cells according to EpCAM expression (NCAM+/CD133−/EpCAM−, NCAM+/CD133−/EpCAMdim, NCAM+/CD133−/EpCAMbright), we show at single-cell resolution a preservation of uninduced and induced cap mesenchyme as well as a transitioning mesenchymal-epithelial state. Concomitantly, differentiating and differentiated epithelial lineages are also maintained. In vitro expansion of discrete stages of early human nephrogenesis in nephron stem cell cultures may be used for drug screening on a full repertoire of developing kidney cells and for prospective isolation of mesenchymal or epithelial renal lineages for regenerative medicine. mNPEM enables in vitro preservation of human renal embryonic CM and epithelia EpCAM allows further dissection of expanded NCAM+CD133− early nephric population Single-cell analysis unveils a continuous lineage hierarchy in nephrogenesis and WT Splice isoform switching confirms a unified MET hierarchy in nephrogenesis and WT
Collapse
Affiliation(s)
- Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; The Dr. Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Rotem Gershon
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Gal Tam
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Itamar Kanter
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Sarit Oriel
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Guy Katz
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; The Dr. Pinchas Borenstein Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer 52621, Israel; The Joseph Buchman Gynecology and Maternity Center, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel
| | - Tomer Kalisky
- Faculty of Engineering and Bar-Ilan Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sheba Centers for Regenerative Medicine and Cancer Research, Sheba Medical Center, Tel-Hashomer 52621, Israel; Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer 52621, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel.
| |
Collapse
|
37
|
Bialucha CU, Collins SD, Li X, Saxena P, Zhang X, Dürr C, Lafont B, Prieur P, Shim Y, Mosher R, Lee D, Ostrom L, Hu T, Bilic S, Rajlic IL, Capka V, Jiang W, Wagner JP, Elliott G, Veloso A, Piel JC, Flaherty MM, Mansfield KG, Meseck EK, Rubic-Schneider T, London AS, Tschantz WR, Kurz M, Nguyen D, Bourret A, Meyer MJ, Faris JE, Janatpour MJ, Chan VW, Yoder NC, Catcott KC, McShea MA, Sun X, Gao H, Williams J, Hofmann F, Engelman JA, Ettenberg SA, Sellers WR, Lees E. Discovery and Optimization of HKT288, a Cadherin-6-Targeting ADC for the Treatment of Ovarian and Renal Cancers. Cancer Discov 2017; 7:1030-1045. [PMID: 28526733 DOI: 10.1158/2159-8290.cd-16-1414] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/11/2017] [Accepted: 05/10/2017] [Indexed: 11/16/2022]
Abstract
Despite an improving therapeutic landscape, significant challenges remain in treating the majority of patients with advanced ovarian or renal cancer. We identified the cell-cell adhesion molecule cadherin-6 (CDH6) as a lineage gene having significant differential expression in ovarian and kidney cancers. HKT288 is an optimized CDH6-targeting DM4-based antibody-drug conjugate (ADC) developed for the treatment of these diseases. Our study provides mechanistic evidence supporting the importance of linker choice for optimal antitumor activity and highlights CDH6 as an antigen for biotherapeutic development. To more robustly predict patient benefit of targeting CDH6, we incorporate a population-based patient-derived xenograft (PDX) clinical trial (PCT) to capture the heterogeneity of response across an unselected cohort of 30 models-a novel preclinical approach in ADC development. HKT288 induces durable tumor regressions of ovarian and renal cancer models in vivo, including 40% of models on the PCT, and features a preclinical safety profile supportive of progression toward clinical evaluation.Significance: We identify CDH6 as a target for biotherapeutics development and demonstrate how an integrated pharmacology strategy that incorporates mechanistic pharmacodynamics and toxicology studies provides a rich dataset for optimizing the therapeutic format. We highlight how a population-based PDX clinical trial and retrospective biomarker analysis can provide correlates of activity and response to guide initial patient selection for first-in-human trials of HKT288. Cancer Discov; 7(9); 1030-45. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.
Collapse
Affiliation(s)
- Carl U Bialucha
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts.
| | - Scott D Collins
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiao Li
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Parmita Saxena
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Xiamei Zhang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Clemens Dürr
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Bruno Lafont
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Pierric Prieur
- Novartis Institutes for Biomedical Research, Novartis Campus, Basel, Switzerland
| | - Yeonju Shim
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Rebecca Mosher
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - David Lee
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Lance Ostrom
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Tiancen Hu
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Sanela Bilic
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | - Vladimir Capka
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Wei Jiang
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Joel P Wagner
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - GiNell Elliott
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Artur Veloso
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jessica C Piel
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Meghan M Flaherty
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Keith G Mansfield
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emily K Meseck
- Novartis Institutes for Biomedical Research, East Hanover, New Jersey
| | - Tina Rubic-Schneider
- Novartis Institutes for Biomedical Research, Campus Klybeckstrasse, Basel, Switzerland
| | | | | | - Markus Kurz
- Novartis Pharma AG, Novartis Campus, Basel, Switzerland
| | - Duc Nguyen
- Novartis Pharma, Cambridge, Massachusetts
| | - Aaron Bourret
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Matthew J Meyer
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Jason E Faris
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Mary J Janatpour
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Vivien W Chan
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | | | | | | | | | - Hui Gao
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Juliet Williams
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Francesco Hofmann
- Novartis Institutes for Biomedical Research, Campus Klybeckstrasse, Basel, Switzerland
| | | | - Seth A Ettenberg
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - William R Sellers
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Emma Lees
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| |
Collapse
|
38
|
Morizane R, Bonventre JV. Kidney Organoids: A Translational Journey. Trends Mol Med 2017; 23:246-263. [PMID: 28188103 DOI: 10.1016/j.molmed.2017.01.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 01/06/2017] [Accepted: 01/08/2017] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cells (hPSCs) are attractive sources for regenerative medicine and disease modeling in vitro. Directed hPSC differentiation approaches have derived from knowledge of cell development in vivo rather than from stochastic cell differentiation. Moreover, there has been great success in the generation of 3D organ-buds termed 'organoids' from hPSCs; these consist of a variety of cell types in vitro that mimic organs in vivo. The organoid bears great potential in the study of human diseases in vitro, especially when combined with CRISPR/Cas9-based genome-editing. We summarize the current literature describing organoid studies with a special focus on kidney organoids, and discuss goals and future opportunities for organoid-based studies.
Collapse
Affiliation(s)
- Ryuji Morizane
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
39
|
Marciano DK. A holey pursuit: lumen formation in the developing kidney. Pediatr Nephrol 2017; 32:7-20. [PMID: 26902755 PMCID: PMC5495142 DOI: 10.1007/s00467-016-3326-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/23/2015] [Accepted: 01/12/2016] [Indexed: 02/07/2023]
Abstract
The formation of polarized epithelial tubules is a hallmark of kidney development. One of the fundamental principles in tubulogenesis is that epithelia coordinate the polarity of individual cells with the surrounding cells and matrix. A central feature in this process is the segregation of membranes into spatially and functionally distinct apical and basolateral domains, and the generation of a luminal space at the apical surface. This review examines our current understanding of the cellular and molecular mechanisms that underlie the establishment of apical-basal polarity and lumen formation in developing renal epithelia, including the roles of cell-cell and cell-matrix interactions and polarity complexes. We highlight growing evidence from animal models, and correlate these findings with models of tubulogenesis from other organ systems, and from in vitro studies.
Collapse
Affiliation(s)
- Denise K. Marciano
- Department of Medicine, Division of Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd. H5.102, Dallas, TX 75390-8856
| |
Collapse
|
40
|
Takasato M, Little MH. A strategy for generating kidney organoids: Recapitulating the development in human pluripotent stem cells. Dev Biol 2016; 420:210-220. [PMID: 27565022 PMCID: PMC6186756 DOI: 10.1016/j.ydbio.2016.08.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/19/2016] [Accepted: 08/21/2016] [Indexed: 02/06/2023]
Abstract
Directed differentiation of human pluripotent stem cells (hPSCs) can provide us any required tissue/cell types by recapitulating the development in vitro. The kidney is one of the most challenging organs to generate from hPSCs as the kidney progenitors are composed of at least 4 different cell types, including nephron, collecting duct, endothelial and interstitium progenitors, that are developmentally distinguished populations. Although the actual developmental process of the kidney during human embryogenesis has not been clarified yet, studies using model animals accumulated knowledge about the origins of kidney progenitors. The implications of these findings for the directed differentiation of hPSCs into the kidney include the mechanism of the intermediate mesoderm specification and its patterning along with anteroposterior axis. Using this knowledge, we previously reported successful generation of hPSCs-derived kidney organoids that contained all renal components and modelled human kidney development in vitro. In this review, we explain the developmental basis of the strategy behind this differentiation protocol and compare strategies of studies that also recently reported the induction of kidney cells from hPSCs. We also discuss the characterization of such kidney organoids and limitations and future applications of this technology.
Collapse
Affiliation(s)
- Minoru Takasato
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; RIKEN Center for Developmental Biology, Kobe 650-0047, Japan.
| | - Melissa H Little
- Murdoch Childrens Research Institute, Parkville, Victoria 3052, Australia; Department of Pediatrics, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
41
|
Abstract
The human kidney develops from four progenitor populations-nephron progenitors, ureteric epithelial progenitors, renal interstitial progenitors and endothelial progenitors-resulting in the formation of maximally 2 million nephrons. Until recently, the reported methods differentiated human pluripotent stem cells (hPSCs) into either nephron progenitor or ureteric epithelial progenitor cells, consequently forming only nephrons or collecting ducts, respectively. Here we detail a protocol that simultaneously induces all four progenitors to generate kidney organoids within which segmented nephrons are connected to collecting ducts and surrounded by renal interstitial cells and an endothelial network. As evidence of functional maturity, proximal tubules within organoids display megalin-mediated and cubilin-mediated endocytosis, and they respond to a nephrotoxicant to undergo apoptosis. This protocol consists of 7 d of monolayer culture for intermediate mesoderm induction, followed by 18 d of 3D culture to facilitate self-organizing renogenic events leading to organoid formation. Personnel experienced in culturing hPSCs are required to conduct this protocol.
Collapse
|
42
|
Gugnoni M, Sancisi V, Gandolfi G, Manzotti G, Ragazzi M, Giordano D, Tamagnini I, Tigano M, Frasoldati A, Piana S, Ciarrocchi A. Cadherin-6 promotes EMT and cancer metastasis by restraining autophagy. Oncogene 2016; 36:667-677. [PMID: 27375021 DOI: 10.1038/onc.2016.237] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 05/25/2016] [Accepted: 06/03/2016] [Indexed: 02/07/2023]
Abstract
The transdifferentiation of epithelial cells toward a mesenchymal condition (EMT) is a complex process that allows tumor cells to migrate to ectopic sites. Cadherins are not just structural proteins, but they act as sensors of the surrounding microenvironment and as signaling centers for cellular pathways. However, the molecular mechanisms underlying these signaling functions remain poorly characterized. Cadherin-6 (CDH6) is a type 2 cadherin, which drives EMT during embryonic development and it is aberrantly re-activated in cancer. We recently showed that CDH6 is a TGFβ target and an EMT marker in thyroid cancer, suggesting a role for this protein in the progression of this type of tumor. Papillary thyroid carcinomas (PTCs) are usually indolent lesions. However, metastatic spreading occurs in about 5% of the cases. The identification of molecular markers that could early predict the metastatic potential of these lesions would be strategic to design more tailored approaches and reduce patients overtreatment. In this work, we assessed the role of CDH6 in the metastatic progression of thyroid cancer. We showed that loss of CDH6 expression profoundly changes cellular architecture, alters the inter-cellular interaction modalities and attenuates EMT features in thyroid cancer cells. Using a yeast two-hybrid screening approach, based on a thyroid cancer patients library, we showed that CDH6 directly interacts with GABARAP, BNIP3 and BNIP3L, and that through these interactions CDH6 restrains autophagy and promotes re-organization of mitochondrial network through a DRP1-mediated mechanism. Analysis of the LIR domains suggests that the interaction with the autophagic machinery may be a common feature of many cadherin family members. Finally, the analysis of CDH6 expression in a unique cohort of human PTCs showed that CDH6 expression marks specifically EMT cells. and it is strongly associated with metastatic behavior and worse outcome of PTCs.
Collapse
Affiliation(s)
- M Gugnoni
- Laboratory of Translational Research, Department of Scientific Direction, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - V Sancisi
- Laboratory of Translational Research, Department of Scientific Direction, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - G Gandolfi
- Laboratory of Translational Research, Department of Scientific Direction, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - G Manzotti
- Laboratory of Translational Research, Department of Scientific Direction, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - M Ragazzi
- Pathology Unit, Deptartment of Oncology and Advanced Technologies, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - D Giordano
- Otolaryngology Unit, Department of General Surgery and Specialistic Unit, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - I Tamagnini
- Pathology Unit, Deptartment of Oncology and Advanced Technologies, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - M Tigano
- Laboratory of Translational Research, Department of Scientific Direction, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - A Frasoldati
- Endocrinology Unit, Department of General Surgery and Specialistic Unit, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - S Piana
- Pathology Unit, Deptartment of Oncology and Advanced Technologies, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - A Ciarrocchi
- Laboratory of Translational Research, Department of Scientific Direction, Arcispedale S Maria Nuova-IRCCS, Reggio Emilia, Italy
| |
Collapse
|
43
|
Zhao L, Jiang R, Xu M, Zhu P, Mo XM, Wang N, Chen GG, Liu ZM. Concomitant high expression of BRAFV600E, P-cadherin and cadherin 6 is associated with High TNM stage and lymph node metastasis in conventional papillary thyroid carcinoma. Clin Endocrinol (Oxf) 2016; 84:748-55. [PMID: 26285159 DOI: 10.1111/cen.12878] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 11/28/2022]
Abstract
CONTEXT AND OBJECTIVE BRAFV600E mutation is the most common activating mutation associated with aggressive behaviours in human tumours including conventional papillary thyroid carcinoma (cPTC). P-cadherin and cadherin 6 have been shown to be mesenchymal-associated cadherins and promote cancer cell invasion and metastasis. The purpose of this study was to examine BRAFV600E, P-cadherin and cadherin 6 expressions in cPTC and to assess the association of their expression with clinicopathological indicators. METHODS BRAFV600E, P-cadherin and cadherin 6 protein expressions in 80 cPTCs, 61 nodular hyperplasia and 76 normal thyroid tissues were examined by immunohistochemistry. The correlation of their protein expression with clinicopathological indicators of cPTC was statistically analysed. RESULTS Protein expression of BRAFV600E, P-cadherin and cadherin 6 was upregulated in cPTC. High protein expression of BRAFV600E, P-cadherin and cadherin 6 was significantly correlated with high TNM stage and lymph node metastasis (LNM) (P < 0·001). Furthermore, BRAFV600E, P-cadherin and cadherin 6 protein expressions were correlated with one another. BRAFV600E high expression combined with both P-cadherin and cadherin-6 high expressions had stronger correlation with high TNM stage and LNM when compared with BRAFV600E high expression combined with either P-cadherin or cadherin-6 high expression (P = 0·042, 0·017 for TNM stage and P = 0·003, 0·006 for LNM, respectively) and only BRAFV600E high expression (P < 0·001 for both TNM stage and LNM). CONCLUSIONS Concomitant high expression of BRAFV600E, P-cadherin and cadherin 6 is strongly associated with high TNM stage and LNM in cPTC.
Collapse
Affiliation(s)
- Le Zhao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Man Xu
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xiao-Mei Mo
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ni Wang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - George G Chen
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin N.T., Hong Kong, China
| | - Zhi-Min Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
44
|
Sharmin S, Taguchi A, Kaku Y, Yoshimura Y, Ohmori T, Sakuma T, Mukoyama M, Yamamoto T, Kurihara H, Nishinakamura R. Human Induced Pluripotent Stem Cell-Derived Podocytes Mature into Vascularized Glomeruli upon Experimental Transplantation. J Am Soc Nephrol 2015; 27:1778-91. [PMID: 26586691 DOI: 10.1681/asn.2015010096] [Citation(s) in RCA: 153] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/26/2015] [Indexed: 01/11/2023] Open
Abstract
Glomerular podocytes express proteins, such as nephrin, that constitute the slit diaphragm, thereby contributing to the filtration process in the kidney. Glomerular development has been analyzed mainly in mice, whereas analysis of human kidney development has been minimal because of limited access to embryonic kidneys. We previously reported the induction of three-dimensional primordial glomeruli from human induced pluripotent stem (iPS) cells. Here, using transcription activator-like effector nuclease-mediated homologous recombination, we generated human iPS cell lines that express green fluorescent protein (GFP) in the NPHS1 locus, which encodes nephrin, and we show that GFP expression facilitated accurate visualization of nephrin-positive podocyte formation in vitro These induced human podocytes exhibited apicobasal polarity, with nephrin proteins accumulated close to the basal domain, and possessed primary processes that were connected with slit diaphragm-like structures. Microarray analysis of sorted iPS cell-derived podocytes identified well conserved marker gene expression previously shown in mouse and human podocytes in vivo Furthermore, we developed a novel transplantation method using spacers that release the tension of host kidney capsules, thereby allowing the effective formation of glomeruli from human iPS cell-derived nephron progenitors. The human glomeruli were vascularized with the host mouse endothelial cells, and iPS cell-derived podocytes with numerous cell processes accumulated around the fenestrated endothelial cells. Therefore, the podocytes generated from iPS cells retain the podocyte-specific molecular and structural features, which will be useful for dissecting human glomerular development and diseases.
Collapse
Affiliation(s)
- Sazia Sharmin
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and
| | - Atsuhiro Taguchi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and
| | - Yusuke Kaku
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and
| | - Yasuhiro Yoshimura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoko Ohmori
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and
| | - Tetsushi Sakuma
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Yamamoto
- Department of Mathematical and Life Sciences, Graduate School of Science, Hiroshima University, Hiroshima, Japan
| | - Hidetake Kurihara
- Division of Anatomy, Juntendo University School of Medicine, Tokyo, Japan; and
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, and Japan Science and Technology Agency, CREST, Kumamoto, Japan
| |
Collapse
|
45
|
Yaoita E, Nishimura H, Nameta M, Yoshida Y, Takimoto H, Fujinaka H, Kawachi H, Magdeldin S, Zhang Y, Xu B, Oyama T, Nakamura F, Yamamoto T. Avian Podocytes, Which Lack Nephrin, Use Adherens Junction Proteins at Intercellular Junctions. J Histochem Cytochem 2015; 64:67-76. [PMID: 26416242 DOI: 10.1369/0022155415611708] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/19/2015] [Indexed: 12/14/2022] Open
Abstract
Nephrin, a major intercellular junction (ICJ) molecule of mammalian podocytes in the renal glomerulus, is absent in the avian genome. We hypothesized that birds use ICJ molecules other than nephrin in their podocytes. Therefore, in the present study, we examined the possible involvement of adherens junction (AJ) proteins in the ICJs of avian podocytes. We found the AJ proteins N-cadherin and α- and β-catenins in podocytes of quail and chickens but not in those of rats, pigs or humans. The AJ proteins were prominent in avian glomerulus-rich fractions in immunoblot analyses, and in immunofluorescence microscopy analyses, they were localized along glomerular capillary walls appearing in at least two staining patterns: weakly diffuse and distinctly granular. Immunoelectron microscopy demonstrated that the significant accumulation of immunogold particles for the AJ proteins were especially evident in avian slit diaphragms and AJs. Furthermore, N-cadherin was found to be expressed in all nephron cells in the early developmental stage but became confined to podocytes during maturation. These results indicate that avian slit diaphragms clearly express AJ proteins as compared with that in the mammal-where AJ proteins are suppressed to an extremely low level-and that avian podocytes are interconnected by AJs per se in addition to slit diaphragms.
Collapse
Affiliation(s)
- Eishin Yaoita
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Hiroko Nishimura
- Department of Health Informatics, Niigata University of Health and Welfare, Niigata, Japan (HN)
| | - Masaaki Nameta
- Cooperative Laboratory of Electron Microscopy, Niigata University, Niigata, Japan (MN)
| | - Yutaka Yoshida
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Hiroki Takimoto
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Hidehiko Fujinaka
- Institute for Clinical Research, Niigata National Hospital, Niigata, Japan (HF)
| | - Hiroshi Kawachi
- Department of Cell Biology (HK), Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Sameh Magdeldin
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Ying Zhang
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Bo Xu
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Tomizo Oyama
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Fujio Nakamura
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| | - Tadashi Yamamoto
- Department of Structural Pathology, Institute of Nephrology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan (EY, YY, HT, SM, YZ, BX, TO, FN, TY)
| |
Collapse
|
46
|
Tanigawa S, Sharma N, Hall MD, Nishinakamura R, Perantoni AO. Preferential Propagation of Competent SIX2+ Nephronic Progenitors by LIF/ROCKi Treatment of the Metanephric Mesenchyme. Stem Cell Reports 2015; 5:435-47. [PMID: 26321142 PMCID: PMC4618653 DOI: 10.1016/j.stemcr.2015.07.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 02/02/2023] Open
Abstract
Understanding the mechanisms responsible for nephrogenic stem cell preservation and commitment is fundamental to harnessing the potential of the metanephric mesenchyme (MM) for nephron regeneration. Accordingly, we established a culture model that preferentially expands the MM SIX2+ progenitor pool using leukemia inhibitory factor (LIF), a Rho kinase inhibitor (ROCKi), and extracellular matrix. Passaged MM cells express the key stem cell regulators Six2 and Pax2 and remain competent to respond to WNT4 induction and form mature tubular epithelia and glomeruli. Mechanistically, LIF activates STAT, which binds to a Stat consensus sequence in the Six2 proximal promoter and sustains SIX2 levels. ROCKi, on the other hand, attenuates the LIF-induced differentiation activity of JNK. Concomitantly, the combination of LIF/ROCKi upregulates Slug expression and activates YAP, which maintains SIX2, PAX2, and SALL1. Using this novel model, our study underscores the pivotal roles of SIX2 and YAP in MM stem cell stability. LIF/ROCKi preserves and selects SIX2+ nephronic progenitors in culture Passaged multipotent progenitors remain competent to respond to inductive cues LIF/ROCKi sustains nuclear levels of SIX2, pPAX2, and YAP
Collapse
Affiliation(s)
- Shunsuke Tanigawa
- National Cancer Institute-Frederick, Frederick, MD 21702, USA; Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan; Program for Leading Graduate Schools, Health Life Science: Interdisciplinary and Glocal (Global and Local) Oriented Program, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | - Nirmala Sharma
- National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | - Michael D Hall
- National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan; Program for Leading Graduate Schools, Health Life Science: Interdisciplinary and Glocal (Global and Local) Oriented Program, Kumamoto University, 2-2-1 Honjo, Kumamoto 860-0811, Japan
| | | |
Collapse
|
47
|
Hirashima T, Adachi T. Procedures for the quantification of whole-tissue immunofluorescence images obtained at single-cell resolution during murine tubular organ development. PLoS One 2015; 10:e0135343. [PMID: 26258587 PMCID: PMC4530862 DOI: 10.1371/journal.pone.0135343] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/21/2015] [Indexed: 11/19/2022] Open
Abstract
Whole-tissue quantification at single-cell resolution has become an inevitable approach for further quantitative understanding of morphogenesis in organ development. The feasibility of the approach has been dramatically increased by recent technological improvements in optical tissue clearing and microscopy. However, the series of procedures required for this approach to lead to successful whole-tissue quantification is far from developed. To provide the appropriate procedure, we here show tips for each critical step of the entire process, including fixation for immunofluorescence, optical clearing, and digital image processing, using developing murine internal organs such as epididymis, kidney, and lung as an example. Through comparison of fixative solutions and of clearing methods, we found optimal conditions to achieve clearer deep-tissue imaging of specific immunolabeled targets and explain what methods result in vivid volume imaging. In addition, we demonstrated that three-dimensional digital image processing after optical clearing produces objective quantitative data for the whole-tissue analysis, focusing on the spatial distribution of mitotic cells in the epididymal tubule. The procedure for the whole-tissue quantification shown in this article should contribute to systematic measurements of cellular processes in developing organs, accelerating the further understanding of morphogenesis at the single cell level.
Collapse
Affiliation(s)
| | - Taiji Adachi
- Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
48
|
Sato S, Kawamata Y, Takahashi A, Imai Y, Hanyu A, Okuma A, Takasugi M, Yamakoshi K, Sorimachi H, Kanda H, Ishikawa Y, Sone S, Nishioka Y, Ohtani N, Hara E. Ablation of the p16(INK4a) tumour suppressor reverses ageing phenotypes of klotho mice. Nat Commun 2015; 6:7035. [PMID: 25923845 PMCID: PMC4421814 DOI: 10.1038/ncomms8035] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 03/26/2015] [Indexed: 01/08/2023] Open
Abstract
The p16INK4a tumour suppressor has an established role in the implementation of cellular senescence in stem/progenitor cells, which is thought to contribute to organismal ageing. However, since p16INK4a knockout mice die prematurely from cancer, whether p16INK4a reduces longevity remains unclear. Here we show that, in mutant mice homozygous for a hypomorphic allele of the α-klotho ageing-suppressor gene (klkl/kl), accelerated ageing phenotypes are rescued by p16INK4a ablation. Surprisingly, this is due to the restoration of α-klotho expression in klkl/kl mice and does not occur when p16INK4a is ablated in α-klotho knockout mice (kl−/−), suggesting that p16INK4a is an upstream regulator of α-klotho expression. Indeed, p16INK4a represses α-klotho promoter activity by blocking the functions of E2Fs. These results, together with the observation that the expression levels of p16INK4a are inversely correlated with those of α-klotho throughout ageing, indicate that p16INK4a plays a previously unrecognized role in downregulating α-klotho expression during ageing. The protein p16INK4a promotes senescence in tissue stem cells and thereby contributes to organismal ageing. Here the authors reveal that p16INK4a also downregulates expression of a-klotho, thereby revealing an additional ageing-promoting function of 16INK4a that is independent from its role in senescence.
Collapse
Affiliation(s)
- Seidai Sato
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan.,Department of Respiratory Medicine and Rheumatology, University of Tokushima Graduate School of Medicine, Tokushima 770-8503, Japan.,CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Yuka Kawamata
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan.,Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan
| | - Akiko Takahashi
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Yoshinori Imai
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Aki Hanyu
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Atsushi Okuma
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Masaki Takasugi
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Kimi Yamakoshi
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Hiroyuki Sorimachi
- Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo 156-8506, Japan
| | - Hiroaki Kanda
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Yuichi Ishikawa
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | - Saburo Sone
- Department of Respiratory Medicine and Rheumatology, University of Tokushima Graduate School of Medicine, Tokushima 770-8503, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, University of Tokushima Graduate School of Medicine, Tokushima 770-8503, Japan
| | - Naoko Ohtani
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan.,Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Noda, Chiba 278-8510, Japan.,PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Eiji Hara
- Division of Cancer Biology, Cancer Institute, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan.,CREST, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan.,Department of Molecular Microbiology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
49
|
Slusser A, Bathula CS, Sens DA, Somji S, Sens MA, Zhou XD, Garrett SH. Cadherin expression, vectorial active transport, and metallothionein isoform 3 mediated EMT/MET responses in cultured primary and immortalized human proximal tubule cells. PLoS One 2015; 10:e0120132. [PMID: 25803827 PMCID: PMC4372585 DOI: 10.1371/journal.pone.0120132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 01/19/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cultures of human proximal tubule cells have been widely utilized to study the role of EMT in renal disease. The goal of this study was to define the role of growth media composition on classic EMT responses, define the expression of E- and N-cadherin, and define the functional epitope of MT-3 that mediates MET in HK-2 cells. METHODS Immunohistochemistry, microdissection, real-time PCR, western blotting, and ELISA were used to define the expression of E- and N-cadherin mRNA and protein in HK-2 and HPT cell cultures. Site-directed mutagenesis, stable transfection, measurement of transepithelial resistance and dome formation were used to define the unique amino acid sequence of MT-3 associated with MET in HK-2 cells. RESULTS It was shown that both E- and N-cadherin mRNA and protein are expressed in the human renal proximal tubule. It was shown, based on the pattern of cadherin expression, connexin expression, vectorial active transport, and transepithelial resistance, that the HK-2 cell line has already undergone many of the early features associated with EMT. It was shown that the unique, six amino acid, C-terminal sequence of MT-3 is required for MT-3 to induce MET in HK-2 cells. CONCLUSIONS The results show that the HK-2 cell line can be an effective model to study later stages in the conversion of the renal epithelial cell to a mesenchymal cell. The HK-2 cell line, transfected with MT-3, may be an effective model to study the process of MET. The study implicates the unique C-terminal sequence of MT-3 in the conversion of HK-2 cells to display an enhanced epithelial phenotype.
Collapse
Affiliation(s)
- Andrea Slusser
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Chandra S. Bathula
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Donald A. Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Seema Somji
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Mary Ann Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Xu Dong Zhou
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
| | - Scott H. Garrett
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, United States of America
- * E-mail:
| |
Collapse
|
50
|
Lindström NO, Carragher NO, Hohenstein P. The PI3K pathway balances self-renewal and differentiation of nephron progenitor cells through β-catenin signaling. Stem Cell Reports 2015; 4:551-60. [PMID: 25754203 PMCID: PMC4400645 DOI: 10.1016/j.stemcr.2015.01.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 01/28/2015] [Accepted: 01/29/2015] [Indexed: 11/24/2022] Open
Abstract
Nephron progenitor cells differentiate to form nephrons during embryonic kidney development. In contrast, self-renewal maintains progenitor numbers and premature depletion leads to impaired kidney function. Here we analyze the PI3K pathway as a point of convergence for the multiple pathways that are known to control self-renewal in the kidney. We demonstrate that a reduction in PI3K signaling triggers premature differentiation of the progenitors and activates a differentiation program that precedes the mesenchymal-to-epithelial transition through ectopic activation of the β-catenin pathway. Therefore, the combined output of PI3K and other pathways fine-tunes the balance between self-renewal and differentiation in nephron progenitors. Nephron progenitor cells require PI3K signaling for self-renewal Reduced PI3K activity enhances β-catenin-induced differentiation Nephron progenitor cells can differentiate prior to completion of epithelialization
Collapse
Affiliation(s)
- Nils Olof Lindström
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XR, UK.
| | - Neil Oliver Carragher
- Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XR, UK
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK; MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK.
| |
Collapse
|